Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Front Cell Neurosci ; 13: 308, 2019.
Article in English | MEDLINE | ID: mdl-31417357

ABSTRACT

Insulin-like growth factor-1 (IGF-1) is a pleiotropic molecule with neurotrophic and immunomodulatory functions. Knowing the capacity of chronically activated microglia to produce IGF-1 may therefore show essential to promote beneficial microglial functions in Alzheimer's disease (AD). Here, we investigated the expression of IGF-1 mRNA and IGF-1 along with the expression of tumor necrosis factor (TNF) mRNA, and the amyloid-ß (Aß) plaque load in the hippocampus of 3- to 24-month-old APPswe/PS1ΔE9 transgenic (Tg) and wild-type (WT) mice. As IGF-1, in particular, is implicated in neurogenesis we also monitored the proliferation of cells in the subgranular zone (sgz) of the dentate gyrus. We found that the Aß plaque load reached its maximum in aged 21- and 24-month-old APPswe/PS1ΔE9 Tg mice, and that microglial reactivity and hippocampal IGF-1 and TNF mRNA levels were significantly elevated in aged APPswe/PS1ΔE9 Tg mice. The sgz cell proliferation decreased with age, regardless of genotype and increased IGF-1/TNF mRNA levels. Interestingly, IGF-1 mRNA was expressed in subsets of sgz cells, likely neuroblasts, and neurons in both genotypes, regardless of age, as well as in glial-like cells. By double in situ hybridization these were shown to be IGF1 mRNA+ CD11b mRNA+ cells, i.e., IGF-1 mRNA-expressing microglia. Quantification showed a 2-fold increase in the number of microglia and IGF-1 mRNA-expressing microglia in the molecular layer of the dentate gyrus in aged APPswe/PS1ΔE9 Tg mice. Double-immunofluorescence showed that IGF-1 was expressed in a subset of Aß plaque-associated CD11b+ microglia and in several subsets of neurons. Exposure of primary murine microglia and BV2 cells to Aß42 did not affect IGF-1 mRNA expression. IGF-1 mRNA levels remained constant in WT mice with aging, unlike TNF mRNA levels which increased with aging. In conclusion, our results suggest that the increased IGF-1 mRNA levels can be ascribed to a larger number of IGF-1 mRNA-expressing microglia in the aged APPswe/PS1ΔE9 Tg mice. The finding that subsets of microglia retain the capacity to express IGF-1 mRNA and IGF-1 in the aged APPswe/PS1ΔE9 Tg mice is encouraging, considering the beneficial therapeutic potential of modulating microglial production of IGF-1 in AD.

2.
Front Cell Neurosci ; 12: 397, 2018.
Article in English | MEDLINE | ID: mdl-30459560

ABSTRACT

Neuroinflammation, characterized by chronic activation of the myeloid-derived microglia, is a hallmark of Alzheimer's disease (AD). Systemic inflammation, typically resulting from infection, has been linked to the progression of AD due to exacerbation of the chronic microglial reaction. However, the mechanism and the consequences of this exacerbation are largely unknown. Here, we mimicked systemic inflammation in AD with weekly intraperitoneal (i.p.) injections of APPSWE/PS1ΔE9 transgenic mice with E. coli lipopolysaccharide (LPS) from 9 to 12 months of age, corresponding to the period with the steepest increase in amyloid pathology. We found that the repeated LPS injections ameliorated amyloid pathology in the neocortex while increasing the neuroinflammatory reaction. To elucidate mechanisms, we analyzed the proteome of the hippocampus from the same mice as well as in unique samples of CNS myeloid cells. The repeated LPS injections stimulated protein pathways of the complement system, retinoid receptor activation and oxidative stress. CNS myeloid cells from transgenic mice showed enrichment in pathways of amyloid-beta clearance and elevated levels of the lysosomal protease cathepsin Z, as well as amyloid precursor protein, apolipoprotein E and clusterin. These proteins were found elevated in the proteome of both LPS and vehicle injected transgenics, and co-localized to CD11b+ microglia in transgenic mice and in primary murine microglia. Additionally, cathepsin Z, amyloid precursor protein, and apolipoprotein E appeared associated with amyloid plaques in neocortex of AD cases. Interestingly, cathepsin Z was expressed in microglial-like cells and co-localized to CD68+ microglial lysosomes in AD cases, and it was expressed in perivascular cells in AD and control cases. Taken together, our results implicate systemic LPS administration in ameliorating amyloid pathology in early-to-mid stage disease in the APPSWE/PS1ΔE9 mouse and attract attention to the potential disease involvement of cathepsin Z expressed in CNS myeloid cells in AD.

3.
Alzheimers Dement (N Y) ; 4: 215-223, 2018.
Article in English | MEDLINE | ID: mdl-29955664

ABSTRACT

INTRODUCTION: Treatment with selective serotonin reuptake inhibitors has been suggested to mitigate amyloid-ß (Aß) pathology in Alzheimer's disease, in addition to an antidepressant mechanism of action. METHODS: We investigated whether chronic treatment with paroxetine, a selective serotonin reuptake inhibitor, mitigates Aß pathology in plaque-bearing double-transgenic amyloid precursor protein (APP)swe/presenilin 1 (PS1)ΔE9 mutants. In addition, we addressed whether serotonin depletion affects Aß pathology. Treatments were assessed by measurement of serotonin transporter occupancy and high-performance liquid chromatography. The effect of paroxetine on Aß pathology was evaluated by stereological plaque load estimation and Aß42/Aß40 ratio by enzyme-linked immunosorbent assay. RESULTS: Contrary to our hypothesis, paroxetine therapy did not mitigate Aß pathology, and depletion of brain serotonin did not exacerbate Aß pathology. However, chronic paroxetine therapy increased mortality in APPswe/PS1ΔE9 transgenic mice. DISCUSSION: Our results question the ability of selective serotonin reuptake inhibitor therapy to ameliorate established Aß pathology. The severe adverse effect of paroxetine may discourage its use for disease-modifying purposes in Alzheimer's disease.

4.
Oncotarget ; 8(36): 60223-60242, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28947966

ABSTRACT

Neuroinflammation is a hallmark of Alzheimer's disease and TNFα as the main inducer of neuroinflammation has neurodegenerative but also pro-regenerative properties, however, the dose-dependent molecular changes on signaling pathway level are not fully understood. We performed quantitative proteomics and phospho-proteomics to target this point. In HT22 cells, we found that TNFα reduced mitochondrial signaling and inhibited mTOR protein translation signaling but also led to induction of neuroprotective MAPK-CREB signaling. Stimulation of human neurons with TNFα revealed similar cellular mechanisms. Moreover, a number of synaptic plasticity-associated genes were altered in their expression profile including CREB. SiRNA-mediated knockdown of CREB in human neurons prior to TNFα stimulation led to a reduced number of protein/phospho-protein hits compared to siRNA-mediated knockdown of CREB or TNFα stimulation alone and countermeasured the reduced CREB signaling. In vivo data of TNFα knockout mice showed that learning ability did not depend on TNFα per se but that TNFα was essential for preserving the learning ability after episodes of lipopolysaccharide-induced neuroinflammation. This may be based on modulation of CREB/CREB signaling as revealed by the in vitro / in vivo data. Our data show that several molecular targets and signaling pathways induced by TNFα in neurons resemble those seen in Alzheimer's disease pathology.

5.
Neurobiol Dis ; 104: 50-60, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28461249

ABSTRACT

Altered neurogenesis may influence hippocampal functions such as learning and memory in Alzheimer's disease. Selective serotonin reuptake inhibitors enhance neurogenesis and have been reported to reduce cerebral amyloidosis in both humans and transgenic mice. We have used stereology to assess the longitudinal changes in the number of doublecortin-expressing neuroblasts and number of granular neurons in the dentate gyrus of APPswe/PS1dE9 transgenic mice. Furthermore, we investigated the effect of long-term paroxetine treatment on the number of neuroblasts and granular neurons, hippocampal amyloidosis, and spontaneous alternation behaviour, a measure of spatial working memory, in transgenic mice. We observed no difference in granular neurons between transgenic and wild type mice up till 18months of age, and no differences with age in wild type mice. The number of neuroblasts and the performance in the spontaneous alternation task was reduced in aged transgenic mice. Paroxetine treatment from 9 to 18months of age reduced hippocampal amyloidosis without affecting the number of neuroblasts or granular neurons. These findings suggest that the amyloidosis affects the differentiation of neuroblasts and spatial working memory, independent of changes in total granular neurons. Furthermore, while long-term paroxetine treatment may be able to reduce hippocampal amyloidosis, it appears to have no effect on total number of granular neurons or spatial working memory.


Subject(s)
Aging/pathology , Alzheimer Disease/pathology , Dentate Gyrus/pathology , Neural Stem Cells/pathology , Neurons/pathology , Aging/genetics , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Bromodeoxyuridine/metabolism , Cytochrome P-450 CYP2D6 Inhibitors/therapeutic use , Dentate Gyrus/drug effects , Disease Models, Animal , Doublecortin Domain Proteins , Exploratory Behavior/drug effects , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Mutation/genetics , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neurogenesis/drug effects , Neurogenesis/physiology , Neurons/drug effects , Neurons/metabolism , Neuropeptides/metabolism , Paroxetine/therapeutic use , Presenilin-1/genetics
6.
Neurobiol Aging ; 36(6): 2164-75, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25892207

ABSTRACT

The susceptibility of the aging brain to neurodegenerative disease may in part be attributed to cellular aging of the microglial cells that survey it. We investigated the effect of cellular aging induced by telomere shortening on microglia by the use of mice lacking the telomerase RNA component (TERC) and design-based stereology. TERC knockout (KO) mice had a significantly reduced number of CD11b(+) microglia in the dentate gyrus. Because of an even greater reduction in dentate gyrus volume, microglial density was, however, increased. Microglia in TERC KO mice maintained a homogenous distribution and normal expression of CD45 and CD68 and the aging marker, ferritin, but were morphologically distinct from microglia in both adult and old wild-type mice. TERC KO mice also showed increased cellular apoptosis and impaired spatial learning. Our results suggest that individual microglia are relatively resistant to telomerase deficiency during steady state conditions, despite an overall reduction in microglial numbers. Furthermore, telomerase deficiency and aging may provide disparate cues leading to distinct changes in microglial morphology and phenotype.


Subject(s)
Aging/genetics , Aging/pathology , Cellular Senescence/genetics , Cellular Senescence/physiology , Microglia/pathology , Phenotype , RNA/genetics , Telomerase/deficiency , Telomerase/genetics , Telomere/pathology , Animals , Antigens, CD , Antigens, Differentiation, Myelomonocytic , Apoptosis/genetics , Dentate Gyrus/cytology , Dentate Gyrus/pathology , Ferritins , Leukocyte Common Antigens , Mice, Inbred C57BL , Mice, Knockout , Neurodegenerative Diseases , Spatial Learning , Telomere/physiology
7.
Brain Behav Immun ; 48: 86-101, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25774009

ABSTRACT

Beta-amyloid (Aß) plaques and chronic neuroinflammation are significant neuropathological features of Alzheimer's disease. Microglial cells in aged brains have potential to produce cytokines such as TNF and IL-1 family members (IL-1α, IL-1ß, and IL-1Ra) and to phagocytose Aß in Alzheimer's disease, however the inter-relationship between these processes is poorly understood. Here we show that % Aß plaque load followed a sigmoidal trajectory with age in the neocortex of APPswe/PS1ΔE9 Tg mice, and correlated positively with soluble Aß40 and Aß42. Aß measures were moderately correlated with mRNA levels of CD11b, TNF, and IL-1Ra. Cytokine production and Aß load were assessed in neocortical CD11b(+)(CD45(+)) microglia by flow cytometry. Whereas most microglia in aged mice produced IL-1Ra, relatively low proportions of microglia produced TNF, IL-1α, and IL-1ß. However, microglial production of these latter cytokines was generally increased in APP/PS1 Tg mice. Microglia that phagocytosed endogenously-produced Aß were only observed in APP/PS1 Tg mice. Differences in phagocytic index and total Aß load were observed in microglia with specific cytokine profiles. Both phagocytic index and total Aß load were higher in IL-1α(+) and IL-1Ra(+) microglia, than microglia that did not produce these cytokines. In contrast, total Aß load was lower in IL-1ß(+) and TNF(+) microglia, compared to IL-1ß(-) and TNF(-) microglia, and TNF(+) microglia also had a lower phagocytic index. Using GFP bone marrow chimeric mice, we confirmed that the majority of neocortical CD11b(+)(CD45(+)) microglia were resident cells (GFP(-)) in APP/PS1 Tg mice, even after selectively analysing CD11b(+)CD45(high) cells, which are typically considered to be infiltrating cells. Together, our data demonstrate that cytokine expression is selectively correlated with age and Aß pathology, and is associated with an altered Aß load in phagocytic microglia from APP/PS1 Tg mice. These findings have implications for understanding the regulation of microglial cytokine production and phagocytosis of Aß in Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/metabolism , Brain/metabolism , Cytokines/metabolism , Microglia/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Disease Models, Animal , Mice , Mice, Transgenic , Presenilin-1/genetics , Presenilin-1/metabolism
8.
Aging Cell ; 13(4): 584-95, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24641683

ABSTRACT

Microglia integrate within the neural tissue with a distinct ramified morphology through which they scan the surrounding neuronal network. Here, we used a digital tool for the quantitative morphometric characterization of fine cortical microglial structures in mice, and the changes they undergo with aging and in Alzheimer's-like disease. We show that, compared with microglia in young mice, microglia in old mice are less ramified and possess fewer branches and fine processes along with a slightly increased proinflammatory cytokine expression. A similar microglial pathology appeared 6-12 months earlier in mouse models of Alzheimer's disease (AD), along with a significant increase in brain parenchyma lacking coverage by microglial processes. We further demonstrate that microglia near amyloid plaques acquire unique activated phenotypes with impaired process complexity. We thus show that along with a chronic proinflammatory reaction in the brain, aging causes a significant reduction in the capacity of microglia to scan their environment. This type of pathology is markedly accelerated in mouse models of AD, resulting in a severe microglial process deficiency, and possibly contributing to enhanced cognitive decline.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Microglia/pathology , Plaque, Amyloid/pathology , Aging/pathology , Alzheimer Disease/metabolism , Animals , Antigens, CD/metabolism , Apyrase/metabolism , CD11b Antigen/metabolism , Cell Shape , Cerebral Cortex/pathology , Cytokines/metabolism , Disease Models, Animal , Green Fluorescent Proteins/metabolism , Inflammation Mediators/metabolism , Leukocyte Common Antigens/metabolism , Mice , Mice, Transgenic , Microglia/metabolism , Plaque, Amyloid/metabolism
9.
Methods Mol Biol ; 1041: 129-45, 2013.
Article in English | MEDLINE | ID: mdl-23813377

ABSTRACT

Microglia are innate immune cells that survey the central nervous system (CNS) and respond almost immediately to any disturbance in CNS homeostasis. They are derived from primitive yolk sac myeloid progenitors and in the mouse colonize the CNS during fetal development. As a population, microglia have the potential to expand rapidly in response to inflammatory stimuli, injury, or any other pathological changes, due to a high capacity for proliferation. In addition, apoptotic mechanisms can be evoked to retract the microglial population, as reactivity declines. In the normal CNS, a low rate of proliferation and apoptosis maintain a low rate of microglial turnover. Here, we describe quantitative analysis of proliferation and apoptosis of microglial cells isolated from individual adult mice by flow cytometry, which allows distinction from perivascular or infiltrating macrophages, based on differential expression of CD45. These methods can be applied to analyze microglial turnover in various models of neuroinflammation.


Subject(s)
Flow Cytometry , Microglia/cytology , Animals , Apoptosis/physiology , Cell Proliferation , Cells, Cultured , Humans
10.
Histol Histopathol ; 26(4): 519-30, 2011 04.
Article in English | MEDLINE | ID: mdl-21360445

ABSTRACT

Microglia are essential cellular components of a well-functioning central nervous system (CNS). The development and establishment of the microglial population differs from the other major cell populations in the CNS i.e. neurons and macroglia (astrocytes and oligodendrocytes). This different ontogeny gives microglia unique properties. In recent years detailed studies of the microglial population have been greatly facilitated by the use of bone marrow (BM) chimeric animals. Experimental BM transplants have provided the opportunity to trace and investigate how BM cells migrate into the CNS and settle to become microglia. Furthermore various functional properties of microglia in the normal and pathological CNS are now being revealed because of combinations of BM transplantations and experimental disease models. Here, we describe some of the latest findings in microglial biology and discuss the potential for using microglia in therapeutic interventions.


Subject(s)
Central Nervous System/cytology , Immune System/cytology , Microglia/cytology , Animals , Apoptosis , Bone Marrow Transplantation , Cell Differentiation , Cell Lineage , Cell Movement , Central Nervous System/embryology , Central Nervous System/immunology , Disease Models, Animal , Humans , Immune System/immunology , Mice , Microglia/immunology , Neurons/pathology , Phagocytosis , Stem Cells/cytology , Stem Cells/immunology , Transplantation Chimera
11.
J Neuropathol Exp Neurol ; 68(8): 845-56, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19606068

ABSTRACT

Acute multiple sclerosis lesions are characterized by accumulation of T cells and macrophages, destruction of myelin and oligodendrocytes, and axonal damage. There is, however, limited information on neuroimmune interactions distal to sites of axonal damage in the T cell-infiltrated central nervous system. We investigated T-cell infiltration, myelin clearance, microglial activation, and phagocytic activity distal to sites of axonal transection through analysis of the perforant pathway deafferented dentate gyrus in SJL mice that had received T cells specific for myelin basic protein (TMBP) or ovalbumin (TOVA). The axonal lesion of TMBP-recipient mice resulted in lesion-specific recruitment of large numbers of T cells in contrast to very limited T-cell infiltration in TOVA-recipient and -naïve perforant pathway-deafferented mice. By double immunofluorescence and confocal microscopy, infiltration with TMBP but not TOVA enhanced the microglial response to axonal transection and microglial phagocytosis of myelin debris associated with the degenerating axons. Because myelin antigen-specific immune responses may provoke protective immunity, increased phagocytosis of myelin debris might enhance regeneration after a neural antigen-specific T cell-mediated immune response in multiple sclerosis.


Subject(s)
Axons/pathology , Central Nervous System/immunology , Microglia/physiology , Myelin Sheath/metabolism , Nerve Degeneration/immunology , T-Lymphocytes/immunology , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , Axotomy/methods , Cell Count/methods , Central Nervous System/metabolism , Central Nervous System/pathology , Female , Macrophage-1 Antigen/metabolism , Mice , Microglia/immunology , Myelin Basic Protein/immunology , Myelin Sheath/immunology , Nerve Degeneration/metabolism , Neurofilament Proteins/metabolism , Perforant Pathway/injuries , Perforant Pathway/pathology , Phagocytes/immunology , Phagocytes/metabolism , Statistics, Nonparametric , T-Lymphocytes/metabolism
12.
J Immunol ; 181(10): 7284-91, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18981150

ABSTRACT

Tissue response to injury includes expression of genes encoding cytokines and chemokines. These regulate entry of immune cells to the injured tissue. The synthesis of many cytokines and chemokines involves NF-kappaB and signal transducers and activators of transcription (STAT). Injury to the CNS induces glial response. Astrocytes are the major glial population in the CNS. We examined expression of STATs and the chemokine CCL2 and their relationship to astroglial NF-kappaB signaling in the CNS following axonal transection. Double labeling with Mac-1/CD11b and glial fibrillary acidic protein revealed that STAT2 up-regulation and phosphorylation colocalized exclusively to astrocytes, suggesting the involvement of STAT2 activating signals selectively in astroglial response to injury. STAT1 was also up-regulated and phosphorylated but not exclusively in astrocytes. Both STAT2 up-regulation and phosphorylation were NF-kappaB -dependent since they did not occur in the lesion-reactive hippocampus of transgenic mice with specific inhibition of NF-kappaB activation in astrocytes. We further showed that lack of NF-kappaB signaling significantly reduced injury-induced CCL2 expression as well as leukocyte infiltration. Our results suggest that NF-kappaB signaling in astrocytes controls expression of both STAT2 and CCL2, and thus regulates infiltration of leukocytes into lesion-reactive hippocampus after axonal injury. Taken together, these findings indicate a central role for astrocytes in directing immune-glial interaction in the CNS injury response.


Subject(s)
Astrocytes/metabolism , Brain Injuries/metabolism , Chemokine CCL2/biosynthesis , NF-kappa B/metabolism , STAT2 Transcription Factor/biosynthesis , Animals , Astrocytes/immunology , Axotomy , Blotting, Western , Brain Injuries/genetics , Brain Injuries/immunology , Chemokine CCL2/genetics , Female , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression , Gene Expression Regulation , Hippocampus/immunology , Hippocampus/injuries , Hippocampus/metabolism , Immunohistochemistry , Mice , Mice, Knockout , NF-kappa B/immunology , Phosphorylation , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , STAT1 Transcription Factor/biosynthesis , STAT1 Transcription Factor/genetics , STAT2 Transcription Factor/genetics
13.
J Immunol ; 181(9): 6481-90, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18941239

ABSTRACT

Injury to the CNS provokes an innate inflammatory reaction that engages infiltrating leukocytes with the capacity to repair and/or exacerbate tissue damage. The initial cues that orchestrate leukocyte entry remain poorly defined. We have used flow cytometry to investigate whether MyD88, an adaptor protein that transmits signals from TLRs and receptors for IL-1 and IL-18, regulates leukocyte infiltration into the stab-injured entorhinal cortex (EC) and into sites of axonal degeneration in the denervated hippocampus. We have previously established the kinetics of leukocyte entry into the denervated hippocampus. We now show that significant leukocyte entry into the EC occurs within 3-12 h of stab injury. Whereas T cells showed small, gradual increases over 8 days, macrophage infiltration was pronounced and peaked within 12-24 h. MyD88 deficiency significantly reduced macrophage and T cell recruitment to the stab-injured EC and the denervated hippocampus at 5 days post-injury. Whereas macrophage and T cell entry remained impaired into the denervated hippocampus of MyD88-deficient mice at 8 days, leukocyte infiltration into the stab-injured EC was restored to levels observed in wild-type mice. Transcripts for TNF-alpha, IL-1beta, and CCL2, which increased >50-fold after stab injury in C57BL/6 mice at the time of peak expression, were severely reduced in injured MyD88 knockout mice. Leukocyte recruitment and gene expression were unaffected in TLR2-deficient or TLR4 mutant mice. No significant differences in gene expression were observed in mice lacking IL-1R or IL-18R. These data show that MyD88-dependent signaling mediates proinflammatory gene expression and leukocyte recruitment after CNS injury.


Subject(s)
Chemotaxis, Leukocyte/immunology , Entorhinal Cortex/immunology , Entorhinal Cortex/pathology , Inflammation Mediators/physiology , Myeloid Differentiation Factor 88/physiology , Signal Transduction/immunology , Animals , Chemotaxis, Leukocyte/genetics , Denervation , Entorhinal Cortex/metabolism , Gene Expression Regulation/immunology , Inflammation Mediators/metabolism , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , Point Mutation , Signal Transduction/genetics , Stereotaxic Techniques , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology
14.
J Neuroinflammation ; 5: 46, 2008 Oct 23.
Article in English | MEDLINE | ID: mdl-18947400

ABSTRACT

BACKGROUND: Interleukin-1beta (IL-1beta) and tumor necrosis factor-alpha (TNF-alpha) are expressed by microglia and infiltrating macrophages following ischemic stroke. Whereas IL-1beta is primarily neurotoxic in ischemic stroke, TNF-alpha may have neurotoxic and/or neuroprotective effects. We investigated whether IL-1beta and TNF-alpha are synthesized by overlapping or segregated populations of cells after ischemic stroke in mice. METHODS: We used flow cytometry and immunohistochemistry to examine cellular co-expression of IL-1beta and TNF-alpha at 6, 12 and 24 hours after permanent middle cerebral artery occlusion in mice, validating the results by the use of bone marrow chimeric mice. RESULTS: We found that IL-1beta and TNF-alpha were expressed in largely segregated populations of CD11b+CD45dim microglia and CD11b+CD45high macrophages, with cells expressing both cytokines only rarely. The number of Gr1+ granulocytes producing IL-1beta or TNF-alpha was very low, and we observed no IL-1beta- or TNF-alpha-expressing T cells or astrocytes. CONCLUSION: Taken together, the results show that IL-1beta and TNF-alpha are produced by largely segregated populations of microglia and macrophages after ischemic stroke in mice. Our findings provide evidence of a functional diversity among different subsets of microglia and macrophages that is potentially relevant to future design of anti-inflammatory therapies in stroke.


Subject(s)
Brain Ischemia/immunology , Encephalitis/immunology , Interleukin-1beta/metabolism , Macrophages/immunology , Microglia/immunology , Tumor Necrosis Factor-alpha/metabolism , Animals , Brain Ischemia/metabolism , Brain Ischemia/physiopathology , CD11 Antigens/immunology , Cell Lineage/immunology , Cells, Cultured , Disease Models, Animal , Encephalitis/metabolism , Encephalitis/physiopathology , Flow Cytometry , Immunohistochemistry , Infarction, Middle Cerebral Artery/immunology , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/physiopathology , Leukocyte Common Antigens/immunology , Macrophages/classification , Male , Mice , Mice, Transgenic , Microglia/classification , Stroke/immunology , Stroke/metabolism , Stroke/physiopathology , Transplantation Chimera
15.
J Neuroinflammation ; 4: 24, 2007 Sep 20.
Article in English | MEDLINE | ID: mdl-17883829

ABSTRACT

BACKGROUND: Matrix metalloproteinases (MMPs) are thought to mediate cellular infiltration in central nervous system (CNS) inflammation by cleaving extracellular matrix proteins associated with the blood-brain barrier. The family of MMPs includes 23 proteinases, including six membrane type-MMPs (MT-MMPs). Leukocyte infiltration is an integral part of the pathogenesis of autoimmune inflammation in the CNS, as occurs in multiple sclerosis and its animal model experimental autoimmune encephalomyelitis (EAE), as well as in the response to brain trauma and injury. We have previously shown that gene expression of the majority of MMPs was upregulated in the spinal cord of SJL mice with severe EAE induced by adoptive transfer of myelin basic protein-reactive T cells, whereas four of the six MT-MMPs (MMP-15, 16, 17 and 24) were downregulated. The two remaining MT-MMPs (MMP-14 and 25) were upregulated in whole tissue. METHODS: We used in vivo models of CNS inflammation and injury to study expression of MT-MMP and cytokine mRNA by real-time RT-PCR. Expression was also assessed in microglia sorted from CNS by flow cytometry, and in primary microglia cultures following treatment with IFNgamma. RESULTS: We now confirm the expression pattern of MT-MMPs in the B6 mouse, independent of effects of adjuvant. We further show expression of all the MT-MMPs, except MMP-24, in microglia. Microglia isolated from mice with severe EAE showed statistically significant downregulation of MMP-15, 17 and 25 and lack of increase in levels of other MT-MMPs. Downregulation of MT-MMPs was also apparent following CNS injury. The pattern of regulation of MT-MMPs in neuroinflammation showed no association with expression of the proinflammatory cytokines TNFalpha, IL-1beta, or IFNgamma. CONCLUSION: CNS inflammation and injury leads to downregulation in expression of the majority of MT-MMPs. Microglia in EAE showed a general downregulation of MT-MMPs, and our findings suggest that MT-MMP levels may inversely correlate with microglial reactivity.


Subject(s)
Central Nervous System Diseases/enzymology , Central Nervous System/pathology , Down-Regulation/physiology , Matrix Metalloproteinase Inhibitors , Animals , Cells, Cultured , Central Nervous System/enzymology , Central Nervous System Diseases/genetics , Encephalomyelitis, Autoimmune, Experimental/enzymology , Encephalomyelitis, Autoimmune, Experimental/genetics , Gene Expression Regulation, Enzymologic/physiology , Inflammation/enzymology , Inflammation/genetics , Matrix Metalloproteinases, Membrane-Associated/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Transgenic
16.
Am J Pathol ; 170(5): 1713-24, 2007 May.
Article in English | MEDLINE | ID: mdl-17456776

ABSTRACT

We have characterized the cellular response to demyelination/remyelination in the central nervous system using the toxin cuprizone, which causes reproducible demyelination in the corpus callosum. Microglia were distinguished from macrophages by relative CD45 expression (CD45(dim)) using flow cytometry. Their expansion occurred rapidly and substantially outnumbered infiltrating macrophages and T cells throughout the course of cuprizone treatment. We used bromodeoxyuridine incorporation and bone marrow chimeras to show that both proliferation and immigration from blood accounted for increased microglial numbers. Microglia adopted an activated phenotype during demyelination, up-regulating major histocompatibility class I and B7.2/CD86. A subpopulation of CD45(dim-high) microglia that expressed reduced levels of CD11b emerged during demyelination. These microglia expressed CD11c and were potent antigen-presenting cells in vitro. T cells were recruited to the demyelinated corpus callosum but did not appear to be activated. Our study highlights the role of microglia as a heterogeneous population of cells in primary demyelination, with the capacity to present antigen, proliferate, and migrate into demyelinated areas.


Subject(s)
Corpus Callosum/immunology , Corpus Callosum/pathology , Demyelinating Diseases/immunology , Animals , Cell Proliferation , Cuprizone/toxicity , Demyelinating Diseases/chemically induced , Demyelinating Diseases/pathology , Female , Flow Cytometry , Immunohistochemistry , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Microglia , Monoamine Oxidase Inhibitors/toxicity , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
17.
J Neurosci ; 26(49): 12826-37, 2006 Dec 06.
Article in English | MEDLINE | ID: mdl-17151286

ABSTRACT

Reactive gliosis is a prominent feature of neurodegenerative and neuroinflammatory disease in the CNS, yet the stimuli that drive this response are not known. There is growing appreciation that signaling through Toll-like receptors (TLRs), which is key to generating innate responses to infection, may have pathogen-independent roles. We show that TLR2 was selectively upregulated by microglia in the denervated zones of the hippocampus in response to stereotactic transection of axons in the entorhinal cortex. In mice lacking TLR2, there were transient, selective reductions in lesion-induced expression of cytokines and chemokines. Recruitment of T cells, but not macrophages, was delayed in TLR2-deficient mice, as well as in mice lacking TNFR1 (tumor necrosis factor receptor 1). TLR2 deficiency also affected microglial proliferative expansion, whereas all of these events were unaffected in TLR4-mutant mice. Consistent with the fact that responses in knock-out mice had all returned to wild-type levels by 8 d, there was no evidence for effects on neuronal plasticity at 20 d. These results identify a role for TLR2 signaling in the early glial response to brain injury, acting as an innate bridge to neuroinflammation.


Subject(s)
Brain Injuries/metabolism , Brain Injuries/pathology , Neurons/metabolism , Neurons/pathology , Signal Transduction/physiology , Toll-Like Receptor 2/physiology , Animals , Axons/pathology , Brain Injuries/genetics , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Microglia/pathology , Signal Transduction/genetics , Toll-Like Receptor 2/biosynthesis , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 2/genetics
18.
Brain Res Brain Res Rev ; 48(2): 178-84, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15850656

ABSTRACT

The distinction between immune-regulatory and effector cytokines and chemokines, and neural growth and survival factors (neurotrophins) becomes increasingly blurred. We discuss here the role of immune cytokines and chemokines as mediators of innate glial responses in the central nervous system. Glial responses to axonal degeneration in the hippocampus dentate gyrus are initiated independently of immune involvement, following transection of afferent entorhinal (perforant path) axons. The glial responses that we measure involve early microglial and somewhat later astrocyte activations. Among the earliest responses are the expression of a wide profile of chemokines, and of the cytokine tumor necrosis factor-alpha (TNFalpha). The cytokine interferon-gamma (IFNgamma) is not normally produced in the CNS, but TNFalpha levels are enhanced if it is present. Viral vector-derived IFNgamma directly induces the expression of chemokines in the CNS, in the absence of any other inflammatory event, but the profiles differ from those induced by axotomy. Chemokines that bind the CCR2 receptor are implicated in traffic of macrophages and T cells to the denervated hippocampus. Innate responses in the immune system are directed by Toll-like receptors (TLR). Our recent studies focus on specific TLR signals as upstream on-switches for glial cytokine and chemokine responses. The biological activity of chemokines is regulated by matrix metalloproteinase enzymes (MMPs) and specific members of this family are expressed in response to axonal lesioning. These findings strengthen the case for the sharing of signals between the immune and nervous system.


Subject(s)
Central Nervous System Diseases/metabolism , Chemokines/metabolism , Cytokines/metabolism , Inflammation/metabolism , Animals , Central Nervous System Diseases/complications , Central Nervous System Diseases/etiology , Cytokines/chemistry , Disease Models, Animal , Humans , Models, Cardiovascular , Neuroglia/physiology , Signal Transduction
19.
J Neurosci ; 23(21): 7922-30, 2003 Aug 27.
Article in English | MEDLINE | ID: mdl-12944523

ABSTRACT

Innate responses in the CNS are critical to first line defense against infection and injury. Leukocytes migrate to inflammatory sites in response to chemokines. We studied leukocyte migration and glial chemokine expression within the denervated hippocampus in response to axonal injury caused by entorhinodentate lesions. A population of Mac1/CD11b+ CD45high macrophages (distinct from CD45low microglia) was specifically detected within the lesion-reactive hippocampus by 12 hr after injury. Significant infiltration by CD3+ T cells did not occur in the denervated hippocampus until 24 hr after axotomy. A broad spectrum of chemokines [RANTES/CCL5, monocyte chemoattractant protein (MCP)-1/CCL2, interferon gamma inducible protein (IP)-10/CXCL10, macrophage inflammatory protein (MIP)-1alpha/CCL3, MIP-1beta/CCL4, and MIP-2/CXCL2] was induced at this time. RANTES/CCL5 was not significantly elevated until 24 hr after axotomy, whereas MCP-1/CCL2 was significantly induced before leukocyte infiltration occurred. Neither T cells nor macrophages infiltrated the denervated hippocampus of CCR2-deficient mice, arguing for a critical role for the CCR2 ligand MCP-1/CCL2 in leukocyte migration. Both T cells and macrophages infiltrated CCR5-deficient hippocampi, showing that CCR5 ligands (including RANTES/CCL5) are not critical to this response. In situ hybridization combined with immunohistochemistry for ionized binding calcium adapter molecule (iba)1 or glial fibrillary acidic protein (GFAP) identified iba1+ microglia and GFAP+ astrocytes as major sources of MCP-1/CCL2 within the lesion-reactive hippocampus. We conclude that leukocyte responses to CNS axonal injury are directed via innate glial production of chemokines.


Subject(s)
Central Nervous System/immunology , Chemokines/biosynthesis , Chemotaxis, Leukocyte , Neuroglia/immunology , Animals , Astrocytes/immunology , Axotomy , Chemokine CCL2/biosynthesis , Chemokine CCL2/genetics , Chemokine CCL5/biosynthesis , Chemokine CCL5/genetics , Chemokines/genetics , Female , Hippocampus/cytology , Hippocampus/immunology , Kinetics , Macrophages/immunology , Mice , Mice, Inbred C57BL , Microglia/immunology , Receptors, CCR2 , Receptors, Chemokine/metabolism , T-Lymphocytes/immunology , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...